Abstract

BackgroundVascular calcification is one of the common complications in diabetes mellitus. Many studies have shown that high glucose (HG) caused cardiovascular calcification, but its underlying mechanism is not fully understood. Recently, medial calcification has been most commonly described in the vessels of patients with diabetes. Chondrocytes were involved in the medial calcification. Recent studies have shown that the conversion into mesenchymal stem cells (MSCs) via the endothelial-to-mesenchymal transition (EndMT) could be triggered in chondrocytes. Our previous research has indicated that HG induced EndMT in human aortic endothelial cells (HAECs). Therefore, we addressed the question of whether HG-induced EndMT could be transitioned into MSCs and differentiated into chondrocytes.MethodsHAECs were divided into three groups: a normal glucose (NG) group, HG group (30 mmol/L), and mannitol (5.5 mmol/L NG + 24.5 mmol/L) group. Pathological changes were investigated using fluorescence microscopy and electron microscopy. Immunofluorescence staining was performed to detect the co-expression of endothelial markers, such as CD31, and fibroblast markers, such as fibroblast-specific protein 1 (FSP-1). The expression of FSP-1 was detected by real time-PCR and western blots. Endothelial-derived MSCs were grown in MSC medium for one week. The expression of the MSCs markers STRO-1, CD44, CD10 and the chondrocyte marker SOX9 was detected by immunofluorescence staining and western blots. Chondrocyte expression was detected by alcian blue staining. Calcium deposits were analyzed by alizarin red staining.ResultsThe incubation of HAECs exposed to HG resulted in a fibroblast-like phenotype. Double staining of the HAECs indicated a co-localization of CD31 and FSP-1. The expression of FSP-1 was significantly increased in the HG group, and the cells undergoing EndMT also expressed STRO-1, CD44 and SOX9 compared with the controls (P < 0.05). Additionally, alcian blue staining in the HG group was positive compared to the NG group. Consistent with the evaluation of SOX9 expression, calcium deposits analyzed by alizarin red staining were also enhanced by the HG treatment. Specifically, we showed that HG-induced EndMT is accompanied by the activation of the canonical Snail pathway.ConclusionsOur study demonstrated that HG could induce endothelial cells transdifferentiation into chondrocyte-like cells via the EndMT, which is mediated in part by the activation of the Snail signaling pathway.

Highlights

  • Vascular calcification is one of the common complications in diabetes mellitus

  • high glucose (HG) induces endothelial-to-mesenchymal transition (EndMT) in human aortic endothelial cells (HAECs) As our previous experiment showed, we found that HAECs treated with 30 mmol/L HG for 48 h induced profound changes, with the cells becoming elongated, spindle-shaped and losing cobblestone morphology under fluorescence microscopy (Figure 1)

  • Immunofluorescence with antibodies for the endothelial marker and the mesenchymal marker demonstrated that the HG-treated cells acquired Fibroblast-specific protein1 (FSP1) staining and lost CD31 staining compared with the control cells (Figure 3)

Read more

Summary

Introduction

Many studies have shown that high glucose (HG) caused cardiovascular calcification, but its underlying mechanism is not fully understood. Medial calcification has been most commonly described in the vessels of patients with diabetes. Recent studies have shown that the conversion into mesenchymal stem cells (MSCs) via the endothelial-to-mesenchymal transition (EndMT) could be triggered in chondrocytes. Cardiovascular complications are the leading cause of death in patients with diabetes mellitus (DM) [1]. Emerging evidence suggests that the presence of vascular calcification in any arterial wall in patients with DM is associated with a 3-4-fold higher risk for mortality and cardiovascular events [2]. Pathological studies have shown that patients with DM exhibit characteristic calcification in the tunica media, which is independently associated with cardiovascular mortality [3]. The mechanisms responsible for these processes in diabetic vascular calcification remain largely unknown

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call